Research Paper Volume 11, Issue 11 pp 3704—3715

Integrating genome-wide association study with regulatory SNP annotation information identified candidate genes and pathways for schizophrenia

Xiao Liang1, *, , Sen Wang1, *, , Li Liu1, , Yanan Du1, , Bolun Cheng1, , Yan Wen1, , Yan Zhao1, , Miao Ding1, , Shiqiang Cheng1, , Mei Ma1, , Lu Zhang1, , Xin Qi1, , Ping Li1, , Xiong Guo1, , Feng Zhang1, ,

  • 1 Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, China
* Equal contribution

Received: March 3, 2019       Accepted: May 29, 2019       Published: June 7, 2019      

https://doi.org/10.18632/aging.102008
How to Cite

Copyright: Liang et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

Background: Schizophrenia is a complex mental disorder. The genetic mechanism of schizophrenia remains elusive now.

Methods: We conducted a large-scale integrative analysis of two genome-wide association studies of schizophrenia with functional annotation datasets of regulatory single-nucleotide polymorphism (rSNP). The significant SNPs identified by the two genome-wide association studies were first annotated to obtain schizophrenia associated rSNPs and their target genes and proteins, respectively. We then compared the integrative analysis results to identify the common rSNPs and their target regulatory genes and proteins, shared by the two genome-wide association studies of schizophrenia. Finally, DAVID tool was used to conduct gene ontology and pathway enrichment analysis of the identified targets genes and proteins.

Results: We detected 53 schizophrenia-associated target genes for rSNP, such as FOS (P value = 2.18×10-20), ATXN1 (P value = 5.22×10-21) and HLA-DQA1 (P value = 1.98×10-10). Pathway enrichment analysis identified 24 pathways for transcription factors binding regions, chromatin interacting regions, long non-coding RNAs, topologically associated domains, circular RNAs and post-translational modifications, such as hsa05034:Alcoholism (P value = 2.57×10-7) and hsa04612:Antigen processing and presentation (P value = 6.82×10-8).

Conclusion: We detected multiple candidate genes, gene ontology terms and pathways for schizophrenia, supporting the functional importance of rSNPs, and providing novel clues for understanding the genetic architecture of schizophrenia.

Introduction

Schizophrenia (SCZ) is a chronic and severe mental disorder, characterized by psychosis, apathy and withdrawal, and cognitive impairment. Depression, anxiety, and substance abuse are additional mental health problems of SCZ condition. The symptoms of SCZ typically appear gradually, start between ages 16 and 30, and never resolve in many cases. About 1% of the population is affected by SCZ during their lifetimes, which is associated with substantial morbidity and mortality, as well as personal and societal costs [1, 2]. Moreover, SCZ is ranked among the top 25 leading causes of disability worldwide [3].

SCZ is a multi-factorial disease, and its occurrence depends on environmental and genetic factors. The high heritability of SCZ points to a major role for inherited genetic variants in the etiology of SCZ, with estimated heritability up to 80% [1, 2]. In recent years, considerable progress has been done in the genetic studies of SCZ [4]. Multiple susceptibility genes have been identified for SCZ [2, 5], such as NRG1 [5], DISC1 [5] and DRD2 [2]. Major histocompatibility complex (MHC) [1] on chromosome 6p is the most significant region for SCZ, which contains many markers reaching genome-wide significance [4]. However, the genetic mechanism of SCZ remains largely unknown.

Genome-wide association studies (GWAS) have great power to identify susceptibility genetic loci associated with complex diseases. Over the years, a number of creditable candidate genes for SCZ has been identified, largely by GWAS [2]. Because a stringent threshold requires a huge sample size to reliably identify risk genes, the significant loci identified by GWAS are usually limited and functionally independent, providing limited information for the mechanism studies of SCZ. Moreover, a large part of genetic variants detected by GWAS are located in non-coding chromosomal regions [4]. It is usually confusing how these non-coding regions implicated would be involved in the development of SCZ.

Functional SNPs located in protein-coding and non-coding genes are named regulatory single nucleotide polymorphisms (rSNPs), which usually have major impacts on gene functions [6]. They mainly include transcription factor binding regions (TFBRs), chromatin interactive regions (CIRs), topologically associated domains (TADs), long non-coding RNAs (lncRNAs) coding regions, and circular RNA (circRNA) regions. Additionally, some SNPs located in the protein coding regions can alter protein post-translational modifications (PTMs) [7], such as phosphorylation, methylation, acetylation, ubiquitination, and glycosylation. The implication of rSNPs in the development of complex diseases has been well documented in previous studies [810]. Integrating GWAS and functional rSNPs annotation information have improved GWAS power and provided novel clues for the genetic studies of complex diseases [1113], such as periodontal diseases [13] and breast cancer [12]. To the best of our knowledge, limited efforts have been paid to explore the functional relevance of rSNPs with SCZ.

In this study, we conducted a large-scale integrative analysis of two GWAS datasets of SCZ with functional annotation datasets of rSNPs. The significant SNPs identified by the two GWAS were first annotated to obtain SCZ-associated rSNPs and their target gene and proteins, respectively. We then compared the integrative analysis results to identify the common rSNPs and their target gene and proteins, shared by the two GWAS of SCZ. Finally, DAVID tool was used to conduct gene ontology (GO) and pathway enrichment analysis of the identified target genes and proteins shared by the two GWAS of SCZ.

Results

Analysis results of GWAS and rSNP annotation datasets

For TFBRs, CIRs, lncRNAs regions, TADs and circRNAs, we identified 1,499 SCZ associated rSNPs, corresponding to 35 genes, such as FOS (P value = 2.18×10-20), GABBR1 (P value = 2.18×10-20), MDK (P value = 1.89×10-10) and ATXN1 (P value = 5.22×10-21). For PTM, we detected 43 rSNPs, corresponding to 18 genes, such as HLA-DQA1 (P value = 1.98×10-10), HLA-DRB1 (P value = 1.36×10-12) and ZSCAN31 (P value = 8.78×10-10) (Table 1).

Table 1. List of SCZ associated rSNPs and their target regulatory genes and proteins shared by both SCZ 1 and SCZ 2.

SNPGeneSNP-related regulatory elementsP value
rs35001169HIST1H3JTFBRs, CIRs, lncRNAs regions, TADs, circRNAs7.40× 10-27
rs35001169HIST1H2AMTFBRs, CIRs, lncRNAs regions, TADs, circRNAs7.40× 10-27
rs35819751MIR3143TFBRs, CIRs, lncRNAs regions, TADs, circRNAs9.52× 10-27
rs66462181HIST1H4ATFBRs, CIRs, lncRNAs regions, TADs, circRNAs1.06× 10-26
rs17695758DNAH8TFBRs, CIRs, lncRNAs regions, TADs, circRNAs1.52× 10-26
rs141342723HIST1H2BLTFBRs, CIRs, lncRNAs regions, TADs, circRNAs1.82× 10-26
rs13209332HIST1H2AKTFBRs, CIRs, lncRNAs regions, TADs, circRNAs2.10× 10-26
rs2232423ZSCAN12PTMs1.71× 10-23
rs33932084PGBD1PTMs5.00× 10-23
rs41266839BTN3A1PTMs4.77× 10-22
rs34788973OR2B2PTMs1.87× 10-21
rs34197618ATXN1TFBRs, CIRs, lncRNAs regions, TADs, circRNAs5.22× 10-21
rs41266779FOSTFBRs, CIRs, lncRNAs regions, TADs, circRNAs2.18× 10-20
rs41266779HIST1H2BKTFBRs, CIRs, lncRNAs regions, TADs, circRNAs2.18× 10-20
rs41266779NUP153TFBRs, CIRs, lncRNAs regions, TADs, circRNAs2.18× 10-20
rs41266779PKHD1TFBRs, CIRs, lncRNAs regions, TADs, circRNAs2.18× 10-20
rs41266779CLIC5TFBRs, CIRs, lncRNAs regions, TADs, circRNAs2.18× 10-20
rs41266779DCDC2TFBRs, CIRs, lncRNAs regions, TADs, circRNAs2.18× 10-20
rs41266779GABBR1TFBRs, CIRs, lncRNAs regions, TADs, circRNAs2.18× 10-20
rs41266779PRIM2TFBRs, CIRs, lncRNAs regions, TADs, circRNAs2.18× 10-20
rs41266779ANKHTFBRs, CIRs, lncRNAs regions, TADs, circRNAs2.18× 10-20
rs41266779HIST1H2AHTFBRs, CIRs, lncRNAs regions, TADs, circRNAs2.18× 10-20
rs35050608MBOAT1TFBRs, CIRs, lncRNAs regions, TADs, circRNAs2.37× 10-20
rs35506517LY86TFBRs, CIRs, lncRNAs regions, TADs, circRNAs3.25× 10-20
rs9393718HIST1H2BJTFBRs, CIRs, lncRNAs regions, TADs, circRNAs3.32× 10-20
rs35555795BTN1A1PTMs9.83× 10-17
rs79780963RNU1-60PTFBRs, CIRs, lncRNAs regions, TADs, circRNAs7.51× 10-16
rs13216828BTN3A2PTMs1.59× 10-15
rs9986596ZKSCAN4PTMs5.51× 10-15
rs3891176HLA-DQB1PTMs3.29× 10-14
rs11693528BMPR2TFBRs, CIRs, lncRNAs regions, TADs, circRNAs5.62× 10-14
rs16891235HIST1H1APTMs1.59× 10-13
rs769949PLCL1TFBRs, CIRs, lncRNAs regions, TADs, circRNAs2.10× 10-13
rs853678ZSCAN31PTMs3.42× 10-13
rs281786MPP4TFBRs, CIRs, lncRNAs regions, TADs, circRNAs3.92× 10-13
rs281786AOX1TFBRs, CIRs, lncRNAs regions, TADs, circRNAs3.92× 10-13
rs35220450RAPH1TFBRs, CIRs, lncRNAs regions, TADs, circRNAs4.05× 10-13
rs35220450INO80DTFBRs, CIRs, lncRNAs regions, TADs, circRNAs4.05× 10-13
rs281760AOX2PTFBRs, CIRs, lncRNAs regions, TADs, circRNAs4.71× 10-13
rs10734901ATP6V0A2TFBRs, CIRs, lncRNAs regions, TADs, circRNAs4.83× 10-13
rs3098341BOLLTFBRs, CIRs, lncRNAs regions, TADs, circRNAs4.89× 10-13
rs10431750KLC1TFBRs, CIRs, lncRNAs regions, TADs, circRNAs5.69× 10-13
rs56155997PDE11ATFBRs, CIRs, lncRNAs regions, TADs, circRNAs5.92× 10-13
rs34525648SLC17A2PTMs1.22× 10-12
rs16822516HLA-DRB1PTMs1.36× 10-12
rs71417869CDC42BPBTFBRs, CIRs, lncRNAs regions, TADs, circRNAs4.02× 10-12
rs2075800HSPA1LPTMs6.00× 10-11
rs35324223MDKTFBRs, CIRs, lncRNAs regions, TADs, circRNAs1.89× 10-10
rs707962HLA-DQA1PTMs1.98× 10-10
rs115817940HLA-DRB5PTMs3.46× 10-10
rs13201753ZKSCAN3PTMs8.78× 10-10
rs950169ADAMTSL3PTMs9.17× 10-10
rs13107325SLC39A8PTMs5.03× 10-9
Note: TFBRs, transcription factor binding regions; CIRs, chromatin interactive regions; TADs, topologically associated domains; PTMs, protein post-translational modifications.

GO enrichment analysis

GO enrichment analysis identified 15 GO terms enriched in the identified target genes of TFBRs, CIRs, lncRNAs, TADs and circRNAs, such as GO:0000786~nucleosome (P value = 1.84×10-10), GO:0046982~protein heterodimerization activity (P value = 5.97×10-7), GO:0000788~nuclear nucleosome (P value = 5.63×10-5) and GO:0006334~nucleosome assembly (P value = 5.70×10-5). For PTMs, we identified 37 SCZ-associated GO terms, such as GO:0002504~antigen processing and presentation of peptide or polysaccharide antigen via MHC class II (P value = 4.79×10-7), GO:0042613~MHC class II protein complex (P value = 1.03×10-6), GO:0042605~peptide antigen binding (P value = 2.26×10-6) and GO:0071556~integral component of lumenal side of endoplasmic reticulum membrane (P value = 2.43×10-6) (Table 2).

Table 2. List of SCZ associated gene ontology terms shared by both SCZ 1 and SCZ 2.

TermTerm descriptionSNP-related regulatory elementsP value
GO:0000786nucleosomeTFBRs, CIRs, lncRNAs regions, TADs, circRNAs1.84× 10-10
GO:0002504Antigen processing and presentation of peptide or polysaccharide antigen via MHC class IIPTMs4.79× 10-7
GO:0046982protein heterodimerization activityTFBRs, CIRs, lncRNAs regions, TADs, circRNAs5.97× 10-7
GO:0042613MHC class II protein complexPTMs1.03× 10-6
GO:0042605Peptide antigen bindingPTMs2.26× 10-6
GO:0071556Integral component of lumenal side of endoplasmic reticulum membranePTMs2.43× 10-6
GO:0030658Transport vesicle membranePTMs5.57× 10-6
GO:0030669Clathrin coated endocytic vesicle membranePTMs7.03× 10-6
GO:0002381Immunoglobulin production involved in immunoglobulin mediated immune responsePTMs8.50× 10-6
GO:0050852T cell receptor signaling pathwayPTMs9.71× 10-6
GO:0012507ER to Golgi transport vesicle membranePTMs1.45× 10-5
GO:0002455Humoral immune response mediated by circulating immunoglobulinPTMs1.78× 10-5
GO:0019882Antigen processing and presentationPTMs1.81× 10-5
GO:0030666Endocytic vesicle membranePTMs2.98× 10-5
GO:0060333Interferon gamma mediated signaling pathwayPTMs3.90× 10-5
GO:0031295T cell costimulationPTMs5.17× 10-5
GO:0000788nuclear nucleosomeTFBRs, CIRs, lncRNAs regions, TADs, circRNAs5.63× 10-5
GO:0006334nucleosome assemblyTFBRs, CIRs, lncRNAs regions, TADs, circRNAs5.70× 10-5
GO:0032588Trans Golgi network membranePTMs5.92× 10-5
GO:0019886Antigen processing and presentation of exogenous peptide antigen via MHC class IIPTMs8.46× 10-5
GO:0032395MHC class II receptor activityPTMs8.78× 10-5
GO:0003677DNA bindingTFBRs, CIRs, lncRNAs regions, TADs, circRNAs2.20× 10-4
GO:0005737cytoplasmTFBRs, CIRs, lncRNAs regions, TADs, circRNAs5.83× 10-4
GO:0043565Sequence specific DNA bindingPTMs1.19× 10-3
GO:0005765Lysosomal membranePTMs1.96× 10-3
GO:2001179Regulation of interleukin 10 secretionPTMs2.86× 10-3
GO:0006342chromatin silencingTFBRs, CIRs, lncRNAs regions, TADs, circRNAs2.91× 10-3
GO:0032673Regulation of interleukin 4 productionPTMs3.81× 10-3
GO:0072643Interferon gamma secretionPTMs6.65× 10-3
GO:0006955Immune responsePTMs6.87× 10-3
GO:0003700Transcription factor activity, sequence specific DNA bindingPTMs1.10× 10-2
GO:0042088T helper 1 type immune responsePTMs1.14× 10-2
GO:0016020MembranePTMs1.17× 10-2
GO:0016045Detection of bacteriumPTMs1.23× 10-2
GO:0002437Inflammatory response to antigenic stimulusPTMs1.42× 10-2
GO:0000139Golgi membranePTMs1.64× 10-2
GO:0031047gene silencing by RNATFBRs, CIRs, lncRNAs regions, TADs, circRNAs1.67× 10-2
GO:0042405nuclear inclusion bodyTFBRs, CIRs, lncRNAs regions, TADs, circRNAs2.19× 10-2
GO:0032689Negative regulation of interferon gamma productionPTMs2.64× 10-2
GO:0035774Positive regulation of insulin secretion involved in cellular response to glucose stimulusPTMs2.73× 10-2
GO:0016021Integral component of membranePTMs2.81× 10-2
GO:0010507Negative regulation of autophagyPTMs3.38× 10-2
GO:0007040Lysosome organizationPTMs3.38× 10-2
GO:0042130Negative regulation of T cell proliferationPTMs3.47× 10-2
GO:0051262Protein tetramerizationPTMs3.75× 10-2
GO:0005654nucleoplasmTFBRs, CIRs, lncRNAs regions, TADs, circRNAs3.77× 10-2
GO:0005887Integral component of plasma membranePTMs3.81× 10-2
GO:0007214gamma-aminobutyric acid signaling pathwayTFBRs, CIRs, lncRNAs regions, TADs, circRNAs3.86× 10-2
GO:0070062extracellular exosomeTFBRs, CIRs, lncRNAs regions, TADs, circRNAs3.98× 10-2
GO:0000790nuclear chromatinTFBRs, CIRs, lncRNAs regions, TADs, circRNAs4.24× 10-2
GO:0002227innate immune response in mucosaTFBRs, CIRs, lncRNAs regions, TADs, circRNAs4.38× 10-2
GO:0032200telomere organizationTFBRs, CIRs, lncRNAs regions, TADs, circRNAs4.72× 10-2
Note: TFBRs, transcription factor binding regions; CIRs, chromatin interactive regions; TADs, topologically associated domains; PTMs, protein post-translational modifications.

Pathway enrichment analysis

For TFBRs, CIRs, lncRNAs, TADs and circRNAs, we identified 3 pathways associated with SCZ, including ha05322:Systemic lupus erythematosus (P value = 3.77×10-8), hsa05034:Alcoholism (P value = 2.57×10-7) and hsa05203:Viral carcinogenesis (P value = 1.78×10-2). For PTMs, we identified 21 pathways associated with SCZ, such as hsa04612:Antigen processing and presentation (P value = 6.82×10-8), hsa05310:Asthma (P value = 7.44×10-7), hsa05332:Graft-versus-host disease (P value = 1.00×10-6) and hsa04672:Intestinal immune network for IgA production (P value = 2.96×10-6) (Table 3).

Table 3. List of SCZ associated pathways shared by both SCZ 1 and SCZ 2.

Term descriptionTerm IDSNP-related regulatory elementsP value
Systemic lupus erythematosushsa05322TFBRs, CIRs, lncRNAs regions, TADs, circRNAs, PTMs3.37× 10-8
Antigen processing and presentationhsa04612PTMs6.82× 10-8
Alcoholismhsa05034TFBRs, CIRs, lncRNAs regions, TADs, circRNAs2.57× 10-7
Toxoplasmosishsa05145PTMs3.06× 10-7
Asthmahsa05310PTMs7.44× 10-7
Graft-versus-host diseasehsa05332PTMs1.00× 10-6
Allograft rejectionhsa05330PTMs1.42× 10-6
Influenza Ahsa05164PTMs1.94× 10-6
Type I diabetes mellitushsa04940PTMs2.10× 10-6
Intestinal immune network for IgA productionhsa04672PTMs2.96× 10-6
Autoimmune thyroid diseasehsa05320PTMs4.03× 10-6
Staphylococcus aureus infectionhsa05150PTMs4.52× 10-6
Viral myocarditishsa05416PTMs5.33× 10-6
Inflammatory bowel disease (IBD)hsa05321PTMs7.58× 10-6
Leishmaniasishsa05140PTMs1.04× 10-5
Rheumatoid arthritishsa05323PTMs1.99× 10-5
Epstein-Barr virus infectionhsa05169PTMs5.30× 10-5
Systemic lupus erythematosushsa05322PTMs7.03× 10-5
Cell adhesion molecules (CAMs)hsa04514PTMs8.36× 10-5
Phagosomehsa04145PTMs9.84× 10-5
Tuberculosishsa05152PTMs1.61× 10-4
Herpes simplex infectionhsa05168PTMs1.78× 10-4
HTLV-I infectionhsa05166PTMs4.71× 10-4
Viral carcinogenesishsa05203TFBRs, CIRs, lncRNAs regions, TADs, circRNAs1.78× 10-2
Note: TFBRs, transcription factor binding regions; CIRs, chromatin interactive regions; TADs, topologically associated domains; PTMs, protein post-translational modifications.

Discussion

Considering that most of SCZ variants identified by GWAS were not causal, integration of the GWAS results with functional rSNPs information is a powerful approach to discover novel candidate genes for SCZ [4]. To evaluate the roles of rSNPs in the pathogenesis of SCZ, we conducted a large-scale integrative genomics analysis of two GWAS datasets of SCZ with functional annotation datasets of rSNPs. We identified multiple candidate genes, GO terms, and biological pathways for SCZ. Our study results support the functional importance of rSNPs in the genetic mechanism of SCZ, and provide novel clues for understanding the genetic architecture of SCZ.

We identified several candidate genes for SCZ, such as FOS, GABBR1, MDK, ATXN1 and ZSCAN31. FOS is classified as one of the immediate early genes (IEGs), which encode not only transcription factors, but also a much wider variety of proteins, including signaling molecules, growth factors and cytoskeletal proteins [14]. Alteration in the expression of IEGs is linked to neuron generation and neuronal cell death. Nadia Cattane et al. have reported that FOS was significantly upregulated in the fibroblasts of SCZ patients [14]. Defects in synaptic plasticity are involved in the pathophysiology of SCZ. Interestingly, SNPs either protective (rs1063169) or positively associated with SCZ (rs7101T) were identified, showing transcription factor c-fos was important in the regulation of synaptic plasticity [15]. Huang et al. observed high FOS expression in non-neural peripheral samples and low FOS expression in the brain tissues of SCZ patients compared with healthy controls, which suggests that FOS is a sensitive marker for SCZ [16]. In addition, detection of FOS in blood samples may be helpful for SCZ diagnosis [16]. These combined results support the functional relevance of FOS with SCZ [1416], which is consistent with our study results.

GABBR1 is another SCZ-associated gene identified by this study. γ-aminobutyric acidB (GABAB) is an inhibitory transmitter molecule acting at neuronal synapses. Functional GABAB receptor requires both the GABBR1 and GABBR2 subunits. GABAB receptor can modulate the release of a number of neurotransmitters, including dopamine, serotonin, noradrenaline, somatostatin, glutamate and GABA [17]. Interestingly, the reduction of GABBR1 in pyramidal cells, and consequent reduction of GABAB receptors, can result in the dysfunction of inhibitory mechanism and increase signal output [18]. Previous studies have also observed association between GABBR1 and SCZ. Fatemi et al. observed a significant reduction of GABBR1 protein in the lateral cerebellum of the subjects with SCZ, bipolar disorder, and major depression [17]. In addition, Zai et al. conducted a case-control study and detected a positive association between GABBR1 and SCZ [19].

Genetic variation in a region on chromosome 11 that contains MDK was significantly associated with SCZ [20]. In addition, MDK also accumulated in senile plaques in the hippocampus of patients with Alzheimer’s disease [20, 21]. Notably, ATXN1 serves as one of the hub genes in the protein-protein interaction network containing many known SCZ risk genes [22]. Actually, ATXN1 is highly expressed in prefrontal cortex, and SCZ patients had significantly decreased ATXN1 expression [22]. In addition, Takeo Saito et al. have revealed an association of rs7779855 in ZSCAN31 with SCZ [23].

GO analysis detected several GO terms for SCZ. One important finding of this study is the disclosure of the MHC class II protein complex (GO:0042613), a class of MHC molecules like human leukocyte antigens HLA-DQA1 and HLA-DQB1, which present antigens to CD4-positive T-lymphocytes. The association between SCZ and the immune system has been repeatedly revealed in genetic, epidemiological and post mortem studies [24]. The protein encoded by HLA-DQA1 gene binds to the protein encoded by HLA-DQB1. Together, they form a functional protein complex called an antigen-binding DQαβ heterodimer, which displays foreign peptides to immune system to trigger body's immune response. Interestingly, HLA alleles were previously shown to be associated with SCZ risk [25]. rs9272105 within HLA-DQA1 explained 1–3% of variation in attentional control, and to a lesser extent, premorbid intelligence quotient in psychotic patients [26]. Additionally, rs9272105 within HLA-DQA1 was also individually associated with variation in neuropsychological function [26]. It has been demonstrated that the level of HLA-DRB1 in SCZ was decreased in peripheral blood samples in contrast with increased level in prefrontal cortex [27]. Moreover, HLA-DPA1 and CD74, which are integral components of the MHC Class II protein complex, were both reduced in hippocampus, amygdala, and dorsolateral prefrontal cortex regions in SCZ [28].

T helper 1 type immune response (GO:0042088) is another significant GO term detected by this study. Delayed hypersensitivity reaction is the classic cell-mediated immune response with sensitized T helper-1 cells. Michael et al. found an attenuated skin reactivity to various antigens in SCZ patients [29]. They also observed significantly diminished responses of schizophrenics to antigen stimulation with tetanus and diptheria antigen presentation [29]. Significantly increased serum level of interleukin-18, a cytokine known to activate T helper 1 type cells in immune responses, has been previously observed in SCZ patients [30].

In addition, we also identified several other GO terms associated with SCZ, such as Golgi membrane (GO:0000139) and GABA signaling pathway (GO:0007214). Previously, it was shown that differentially expressed genes related to Golgi apparatus, vesicular transport and membrane association were over-represented in SCZ [31]. In line, Devor et al. identified a large number of GABA-associated genes for SCZ [32].

The involvement of cell adhesion molecules (CAMs) in the pathophysiology of SCZ has long been hypothesized. In this study, CAMs (hsa04514) pathway was detected for SCZ. The CAMs pathway is implicated in a variety of neurocognitive processes, including memory and attention-related reaction time. Multiple CAM genes has been reported to be associated with SCZ risk [26]. Neural CAMs are very important members of the exclusive group of the molecules responsible for precise wiring of nervous system. Neural CAMs serve as ‘‘glue’’ in cell-to-cell adhesion and contact-mediated attraction [33]. They can interact with numerous matrix components, and are involved in many aspects of neuronal development, synaptogenesis, and myelination, which guide brain morphology and support highly-coordinated brain activity [33]. Cerebrospinal fluid neuronal CAMs were significantly increased in SCZ patients [34]. Additionally, the plasma levels of intercellular adhesion molecule-1 was also elevated in SCZ patients [35]. Honer et al. found that syntaxin immunoreactivity in the cingulate cortex from schizophrenics was increased, along with neural CAMs and the CAMs to synaptophysin ratio [36]. Besides, it has been reported that L1 cell adhesion molecule interaction was involved in neuronal function [37].

Another interesting SCZ associated pathway is alcoholism (hsa05034). Recent studies have suggested that alcoholism has a wide-reaching influence on the clinical course of SCZ, contributing to shape abnormalities in hippocampus and subcortical shape differences [38]. We also observed that systemic lupus erythematosus (SLE) (hsa05322) was associated with SCZ. Despite the fact that SCZ is not classified as a typical autoimmune diseases, the dysregulation of the immune system cannot be excluded [39, 40]. Interestingly, DNA and myelin basic protein (MBP)-hydrolyzing antibodies, which play an important harmful role in SLE pathogenesis, were also detected in the sera of SCZ patients. In addition, light chains of IgGs from SCZ patients were similar to those of SLE patients [41].

The majority of SNPs identified by GWAS are enriched in non-coding regions [4], and contribute to complex traits and diseases through various molecular mechanisms. These include effects on transcription factor binding affinities, which can result in differential gene expression [11]. However, significant loci identified by GWAS have rarely been tracked to causal polymorphisms thus far. Integrative analysis of GWAS with functional rSNPs is helpful to improve GWAS power and provide novel clues for pathogenetic studies of SCZ. Notably, the functional SNPs data can assist to exclude unlikely genes/loci, effectively reducing the number of tests needed for unbiased searches across the genome, thus, improving the power to discover novel causal loci [4]. To the best of our knowledge, this is the first large-scale integrative analysis of GWAS and rSNPs for SCZ. The implication of rSNPs in the development of SCZ was systematically investigated considering TFBRs, CIRs, lncRNAs regions, TADs, circRNAs, and PTMs in our study. Nevertheless, one limitation of this study should be noted. The SCZ-associated SNP sets were driven from previous GWAS. The accuracy of our integrative analysis may be affected by the power of previous GWAS of SCZ. Therefore, further studies are warranted to confirm our findings.

In conclusion, we conducted a large-scale integrative genomics analysis of two GWAS datasets of SCZ with functional annotation datasets of rSNPs to explore the genetic basis of rSNPs in the pathogenesis of SCZ. We observed multiple candidate genes, GO terms and pathways for SCZ. We hope that our study results could provide novel clues for the pathogenic and therapic studies of SCZ.

Materials and Methods

The first GWAS dataset of SCZ (SCZ1)

A large GWAS meta-analysis data of SCZ was driven from the Psychiatric Genomics Consortium (PGC) [42], totally containing 33,426 SCZ cases and 32,541 controls. Genotypes from all studies were processed by the PGC using unified quality control procedures followed by imputation of SNPs and insertion-deletions using the 1000 Genomes Project reference panel [43]. Quality control and imputation were performed on each of the study cohort datasets, according to the standard procedures established by the PGC [42]. Genotype imputation was performed using the pre-phasing/imputation stepwise approach implemented in IMPUTE2 [44] and SHAPEIT [45]. The imputation reference set consists of 2,186 phased haplotypes from the full 1000 Genomes Project dataset. Logistic regression was conducted to control for 13 components of ancestry, study sites and genotyping platform. Detailed description of sample characteristics, experimental design, statistical analysis and quality control can be found in the previous studies [42].

The second GWAS dataset of SCZ (SCZ2)

Another independent GWAS data of SCZ [2] was used here. Briefly, this GWAS included 36,989 SCZ cases and 113,075 controls, from 49 ancestry matched, non-overlapping case-control samples (46 of European and three of East Asian ancestry, 34,241 cases and 45,604 controls) and 3 family-based samples of European ancestry (1,235 parent affected-offspring trios). Genotypes from all studies were processed by the PGC using unified quality control procedures. The 1000 Genomes Project reference panel was used for SNPs imputation [43]. In each sample, association testing was conducted using imputed SNP dosages and principal components to control for population stratification. After quality control (imputation INFO score ≥ 0.6, MAF ≥ 0.01, and successfully imputed in ≥ 20 samples), they considered around 9.5 million variants. An inverse-weighted fixed effects model was used for final meta-analysis. Detailed description of sample characteristics, experimental design, statistical analysis and quality control can be found in the previous study [2].

rSNPs annotation datasets

The rSNPs annotation information were driven from the rSNPBase 3.1 database (http://rsnp3.psych.ac.cn) [46] and the AWESOME database (http://www.awesome-hust.com) [7]. rSNPBase 3.1 provided rich functional annotation for human SNP-related regulatory elements and their target regulatory genes, including TFBRs, CIRs, mature microRNA (miRNA) regions, predicted miRNA target sites, lncRNA regions, TADs and circRNAs. AWESOME database is an analysis tool that systematically evaluates the role of SNPs on nearly all kinds of PTMs based on 20 available tools. They construct a comprehensive platform to collect and integrate SNPs and multiple PTM information, utilizing 24 published database or tools. 1,043,608 germline missense variants from the dbSNP was used and each SNP was matched with its protein sequence in AWESOME. Detailed description of the two rSNPs annotation database can be found in the published studies [7, 46].

Statistical analysis

The significant SNPs with GWAS P value < 5.0 × 10-8 were selected from the two GWAS of SCZ (SCZ1 and SCZ2). The selected SCZ-associated SNPs were then annotated by the rSNPBase 3.1 database [46] and the AWESOME database to obtain SCZ associated rSNPs and their target regulatory genes and proteins. We then compared the integrative analysis results to identify the common rSNPs and their target genes and proteins shared by the two GWAS of SCZ. To explore the functional relevance of identified target regulatory genes and proteins with SCZ, GO and pathway enrichment analyses of the identified common target genes and proteins shared by the SCZ1 and SCZ2 were performed by the Database for Annotation, Visualization and Integrated Discovery (DAVID) tool [47].

Abbreviations

GWAS: genome wide association studies; rSNP: regulatory SNP; TFBRs: transcription factor binding regions; CIRs: chromatin interactive regions; lncRNAs: long non-coding RNA regions; TADs: topologically associated domains; circRNAs: circular RNAs; PTMs: protein post-translational modifications; GO: gene ontology; SCZ: Schizophrenia.

Conflicts of Interest

None.

Funding

This work was supported by the National Natural Scientific Foundation of China (81673112); the Key projects of international cooperation among governments in scientific and technological innovation (2016YFE0119100); the Natural Science Basic Research Plan in Shaanxi Province of China (2017JZ024); and the Fundamental Research Funds for the Central Universities.

References

  • 1. Purcell SM, Wray NR, Stone JL, Visscher PM, O’Donovan MC, Sullivan PF, Sklar P, and International Schizophrenia Consortium. Common polygenic variation contributes to risk of schizophrenia and bipolar disorder. Nature. 2009; 460:748–52. https://doi.org/10.1038/nature08185 [PubMed]
  • 2. Schizophrenia Working Group of the Psychiatric Genomics Consortium. Biological insights from 108 schizophrenia-associated genetic loci. Nature. 2014; 511:421–27. https://doi.org/10.1038/nature13595 [PubMed]
  • 3. Chong HY, Teoh SL, Wu DB, Kotirum S, Chiou CF, Chaiyakunapruk N. Global economic burden of schizophrenia: a systematic review. Neuropsychiatr Dis Treat. 2016; 12:357–73. https://doi.org/10.2147/ndt.s96649 [PubMed]
  • 4. Chen J, Cao F, Liu L, Wang L, Chen X. Genetic studies of schizophrenia: an update. Neurosci Bull. 2015; 31:87–98. https://doi.org/10.1007/s12264-014-1494-4 [PubMed]
  • 5. Craddock N, O’Donovan MC, Owen MJ. Genes for schizophrenia and bipolar disorder? Implications for psychiatric nosology. Schizophr Bull. 2006; 32:9–16. https://doi.org/10.1093/schbul/sbj033 [PubMed]
  • 6. Ramírez-Bello J, Jiménez-Morales M. [Functional implications of single nucleotide polymorphisms (SNPs) in protein-coding and non-coding RNA genes in multifactorial diseases]. Gac Med Mex. 2017; 153:238–50. https://doi.org/10.1186/1471-2156-9-10 [PubMed]
  • 7. Yang Y, Peng X, Ying P, Tian J, Li J, Ke J, Zhu Y, Gong Y, Zou D, Yang N, Wang X, Mei S, Zhong R, et al. AWESOME: a database of SNPs that affect protein post-translational modifications. Nucleic Acids Res. 2019; 47:D874–D880. https://doi.org/10.1093/nar/gky821 [PubMed]
  • 8. Prokunina L, Alarcón-Riquelme ME. Regulatory SNPs in complex diseases: their identification and functional validation. Expert Rev Mol Med. 2004; 6:1–15. https://doi.org/10.1017/S1462399404007690 [PubMed]
  • 9. Butler MG, McGuire AB, Masoud H, Manzardo AM. Currently recognized genes for schizophrenia: high-resolution chromosome ideogram representation. Am J Med Genet B Neuropsychiatr Genet. 2016; 171B:181–202. https://doi.org/10.1002/ajmg.b.32391 [PubMed]
  • 10. Wong KC. DNA Motif Recognition Modeling from Protein Sequences. iScience. 2018; 7:198–211. https://doi.org/10.1016/j.isci.2018.09.003 [PubMed]
  • 11. Cavalli M, Pan G, Nord H, Wallén Arzt E, Wallerman O, Wadelius C. Allele-specific transcription factor binding in liver and cervix cells unveils many likely drivers of GWAS signals. Genomics. 2016; 107:248–54. https://doi.org/10.1016/j.ygeno.2016.04.006 [PubMed]
  • 12. Liu Y, Walavalkar NM. Identification of breast cancer associated variants that modulate transcription factor binding. PLoS Genet. 2017; 13:e1006761. https://doi.org/10.1371/journal.pgen.1006761 [PubMed]
  • 13. Razzouk S. Regulatory elements and genetic variations in periodontal diseases. Arch Oral Biol. 2016; 72:106–15. https://doi.org/10.1016/j.archoralbio.2016.08.015 [PubMed]
  • 14. Cattane N, Minelli A, Milanesi E, Maj C, Bignotti S, Bortolomasi M, Bocchio Chiavetto L, Gennarelli M. Altered gene expression in schizophrenia: findings from transcriptional signatures in fibroblasts and blood. PLoS One. 2015; 10:e0116686. https://doi.org/10.1371/journal.pone.0116686 [PubMed]
  • 15. Boyajyan A, Zakharyan R, Atshemyan S, Chavushyan A, Mkrtchyan G. Schizophrenia-associated Risk and Protective Variants of c-Fos Encoding Gene. Recent Adv DNA Gene Seq. 2015; 9:51–57. https://doi.org/10.2174/2352092209666150223113334 [PubMed]
  • 16. Huang J, Liu F, Wang B, Tang H, Teng Z, Li L, Qiu Y, Wu H, Chen J. Central and Peripheral Changes in FOS Expression in Schizophrenia Based on Genome-Wide Gene Expression. Front Genet. 2019; 10:232. https://doi.org/10.3389/fgene.2019.00232 [PubMed]
  • 17. Fatemi SH, Folsom TD, Thuras PD. Deficits in GABA(B) receptor system in schizophrenia and mood disorders: a postmortem study. Schizophr Res. 2011; 128:37–43. https://doi.org/10.1016/j.schres.2010.12.025 [PubMed]
  • 18. Mizukami K, Ishikawa M, Hidaka S, Iwakiri M, Sasaki M, Iritani S. Immunohistochemical localization of GABAB receptor in the entorhinal cortex and inferior temporal cortex of schizophrenic brain. Prog Neuropsychopharmacol Biol Psychiatry. 2002; 26:393–96. https://doi.org/10.1016/S0278-5846(01)00247-0 [PubMed]
  • 19. Zai G, King N, Wong GW, Barr CL, Kennedy JL. Possible association between the gamma-aminobutyric acid type B receptor 1 (GABBR1) gene and schizophrenia. Eur Neuropsychopharmacol. 2005; 15:347–52. https://doi.org/10.1016/j.euroneuro.2004.12.006 [PubMed]
  • 20. Rietschel M, Mattheisen M, Degenhardt F, Mühleisen TW, Kirsch P, Esslinger C, Herms S, Demontis D, Steffens M, Strohmaier J, Haenisch B, Breuer R, Czerski PM, et al, and Genetic Risk and Outcome in Psychosis (GROUP Investigators), and SGENE-plus Consortium. Association between genetic variation in a region on chromosome 11 and schizophrenia in large samples from Europe. Mol Psychiatry. 2012; 17:906–17. https://doi.org/10.1038/mp.2011.80 [PubMed]
  • 21. Yasuhara O, Muramatsu H, Kim SU, Muramatsu T, Maruta H, McGeer PL. Midkine, a novel neurotrophic factor, is present in senile plaques of Alzheimer disease. Biochem Biophys Res Commun. 1993; 192:246–51. https://doi.org/10.1006/bbrc.1993.1406 [PubMed]
  • 22. Liu J, Su B. Integrated analysis supports ATXN1 as a schizophrenia risk gene. Schizophr Res. 2018; 195:298–305. https://doi.org/10.1016/j.schres.2017.10.010 [PubMed]
  • 23. Saito T, Kondo K, Iwayama Y, Shimasaki A, Aleksic B, Yamada K, Toyota T, Hattori E, Esaki K, Ujike H, Inada T, Kunugi H, Kato T, et al. Replication and cross-phenotype study based upon schizophrenia GWASs data in the Japanese population: support for association of MHC region with psychosis. Am J Med Genet B Neuropsychiatr Genet. 2014; 165B:421–27. https://doi.org/10.1002/ajmg.b.32246 [PubMed]
  • 24. Ormel PR, van Mierlo HC, Litjens M, Strien MEV, Hol EM, Kahn RS, de Witte LD. Characterization of macrophages from schizophrenia patients. NPJ Schizophr. 2017; 3:41. https://doi.org/10.1038/s41537-017-0042-4 [PubMed]
  • 25. Andreassen OA, Harbo HF, Wang Y, Thompson WK, Schork AJ, Mattingsdal M, Zuber V, Bettella F, Ripke S, Kelsoe JR, Kendler KS, O’Donovan MC, Sklar P, et al, and Psychiatric Genomics Consortium (PGC) Bipolar Disorder and Schizophrenia Work Groups, and International Multiple Sclerosis Genetics Consortium (IMSGC). Genetic pleiotropy between multiple sclerosis and schizophrenia but not bipolar disorder: differential involvement of immune-related gene loci. Mol Psychiatry. 2015; 20:207–14. https://doi.org/10.1038/mp.2013.195 [PubMed]
  • 26. Hargreaves A, Anney R, O’Dushlaine C, Nicodemus KK, Gill M, Corvin A, Morris D, Donohoe G, and Schizophrenia Psychiatric Genome-Wide Association Study Consortium (PGC-SCZ), and Wellcome Trust Case Control Consortium 2. The one and the many: effects of the cell adhesion molecule pathway on neuropsychological function in psychosis. Psychol Med. 2014; 44:2177–87. https://doi.org/10.1017/S0033291713002663 [PubMed]
  • 27. Mohammadi A, Rashidi E, Amooeian VG. Brain, blood, cerebrospinal fluid, and serum biomarkers in schizophrenia. Psychiatry Res. 2018; 265:25–38. https://doi.org/10.1016/j.psychres.2018.04.036 [PubMed]
  • 28. Morgan LZ, Rollins B, Sequeira A, Byerley W, DeLisi LE, Schatzberg AF, Barchas JD, Myers RM, Watson SJ, Akil H, Bunney WEJr, Vawter MP. Quantitative Trait Locus and Brain Expression of HLA-DPA1 Offers Evidence of Shared Immune Alterations in Psychiatric Disorders. Microarrays (Basel). 2016; 5:6. https://doi.org/10.3390/microarrays5010006 [PubMed]
  • 29. Riedel M, Spellmann I, Schwarz MJ, Strassnig M, Sikorski C, Möller HJ, Müller N. Decreased T cellular immune response in schizophrenic patients. J Psychiatr Res. 2007; 41:3–7. https://doi.org/10.1016/j.jpsychires.2005.11.007 [PubMed]
  • 30. Tanaka KF, Shintani F, Fujii Y, Yagi G, Asai M. Serum interleukin-18 levels are elevated in schizophrenia. Psychiatry Res. 2000; 96:75–80. https://doi.org/10.1016/S0165-1781(00)00196-7 [PubMed]
  • 31. Mudge J, Miller NA, Khrebtukova I, Lindquist IE, May GD, Huntley JJ, Luo S, Zhang L, van Velkinburgh JC, Farmer AD, Lewis S, Beavis WD, Schilkey FD, et al. Genomic convergence analysis of schizophrenia: mRNA sequencing reveals altered synaptic vesicular transport in post-mortem cerebellum. PLoS One. 2008; 3:e3625. https://doi.org/10.1371/journal.pone.0003625 [PubMed]
  • 32. Devor A, Andreassen OA, Wang Y, Mäki-Marttunen T, Smeland OB, Fan CC, Schork AJ, Holland D, Thompson WK, Witoelar A, Chen CH, Desikan RS, McEvoy LK, et al. Genetic evidence for role of integration of fast and slow neurotransmission in schizophrenia. Mol Psychiatry. 2017; 22:792–801. https://doi.org/10.1038/mp.2017.33 [PubMed]
  • 33. Gnanapavan S, Giovannoni G. Neural cell adhesion molecules in brain plasticity and disease. Mult Scler Relat Disord. 2013; 2:13–20. https://doi.org/10.1016/j.msard.2012.08.002 [PubMed]
  • 34. van Kammen DP, Poltorak M, Kelley ME, Yao JK, Gurklis JA, Peters JL, Hemperly JJ, Wright RD, Freed WJ. Further studies of elevated cerebrospinal fluid neuronal cell adhesion molecule in schizophrenia. Biol Psychiatry. 1998; 43:680–86. https://doi.org/10.1016/S0006-3223(97)00324-7 [PubMed]
  • 35. Nguyen TT, Dev SI, Chen G, Liou SC, Martin AS, Irwin MR, Carroll JE, Tu X, Jeste DV, Eyler LT. Abnormal levels of vascular endothelial biomarkers in schizophrenia. Eur Arch Psychiatry Clin Neurosci. 2018; 268:849–60. https://doi.org/10.1007/s00406-017-0842-6 [PubMed]
  • 36. Honer WG, Falkai P, Young C, Wang T, Xie J, Bonner J, Hu L, Boulianne GL, Luo Z, Trimble WS. Cingulate cortex synaptic terminal proteins and neural cell adhesion molecule in schizophrenia. Neuroscience. 1997; 78:99–110. https://doi.org/10.1016/S0306-4522(96)00489-7 [PubMed]
  • 37. Aberg KA, Liu Y, Bukszár J, McClay JL, Khachane AN, Andreassen OA, Blackwood D, Corvin A, Djurovic S, Gurling H, Ophoff R, Pato CN, Pato MT, et al. A comprehensive family-based replication study of schizophrenia genes. JAMA Psychiatry. 2013; 70:573–81. https://doi.org/10.1001/jamapsychiatry.2013.288 [PubMed]
  • 38. Smith MJ, Wang L, Cronenwett W, Goldman MB, Mamah D, Barch DM, Csernansky JG. Alcohol use disorders contribute to hippocampal and subcortical shape differences in schizophrenia. Schizophr Res. 2011; 131:174–83. https://doi.org/10.1016/j.schres.2011.05.014 [PubMed]
  • 39. Bergink V, Gibney SM, Drexhage HA. Autoimmunity, inflammation, and psychosis: a search for peripheral markers. Biol Psychiatry. 2014; 75:324–31. https://doi.org/10.1016/j.biopsych.2013.09.037 [PubMed]
  • 40. Strous RD, Shoenfeld Y. Schizophrenia, autoimmunity and immune system dysregulation: a comprehensive model updated and revisited. J Autoimmun. 2006; 27:71–80. https://doi.org/10.1016/j.jaut.2006.07.006 [PubMed]
  • 41. Ermakov EA, Ivanova SA, Buneva VN, Nevinsky GA. Hydrolysis by catalytic IgGs of microRNA specific for patients with schizophrenia. IUBMB Life. 2018; 70: 153–164. https://doi.org/10.1002/iub.1712 [PubMed]
  • 42. Ruderfer DM, Ripke S, McQuillin A, Boocock J, Stahl EA, Pavlides JM, Mullins N, Charney AW, Ori AP, Loohuis LM, Domenici E, Di Florio A, Papiol S, et al, and Bipolar Disorder and Schizophrenia Working Group of the Psychiatric Genomics Consortium. Electronic address: douglas.ruderfer@vanderbilt.edu, and Bipolar Disorder and Schizophrenia Working Group of the Psychiatric Genomics Consortium. Genomic Dissection of Bipolar Disorder and Schizophrenia, Including 28 Subphenotypes. Cell. 2018; 173:1705–1715.e16. https://doi.org/10.1016/j.cell.2018.05.046 [PubMed]
  • 43. Mathelier A, Shi W, Wasserman WW. Identification of altered cis-regulatory elements in human disease. Trends Genet. 2015; 31:67–76. https://doi.org/10.1016/j.tig.2014.12.003 [PubMed]
  • 44. Howie B, Marchini J, Stephens M. Genotype imputation with thousands of genomes. G3 (Bethesda). 2011; 1:457–70. https://doi.org/10.1534/g3.111.001198 [PubMed]
  • 45. Delaneau O, Zagury JF, Marchini J. Improved whole-chromosome phasing for disease and population genetic studies. Nat Methods. 2013; 10:5–6. https://doi.org/10.1038/nmeth.2307 [PubMed]
  • 46. Guo L, Wang J. rSNPBase 3.0: an updated database of SNP-related regulatory elements, element-gene pairs and SNP-based gene regulatory networks. Nucleic Acids Res. 2018; 46:D1111–16. https://doi.org/10.1093/nar/gkx1101 [PubMed]
  • 47. Li T, Underhill J, Liu XH, Sham PC, Donaldson P, Murray RM, Wright P, Collier DA. Transmission disequilibrium analysis of HLA class II DRB1, DQA1, DQB1 and DPB1 polymorphisms in schizophrenia using family trios from a Han Chinese population. Schizophr Res. 2001; 49:73–78. https://doi.org/10.1016/S0920-9964(00)00111-0 [PubMed]